Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Bioelectromagnetics ; 35(8): 559-67, 2014 Dec.
Article in English | MEDLINE | ID: mdl-25256206

ABSTRACT

Studies describing the influence of radiofrequency electromagnetic fields on bone marrow cells (BMC) often lack functional data. We examined the effects of in vivo exposure to a Global System for Mobile Communications (GSM) modulated 900 MHz RF fields on BMC using two transplantation models. X-irradiated syngeneic mice were injected with BMC from either RF-field-exposed, sham-exposed or cage control mice. Twelve weeks after transplantation, no differences in thymocyte number, frequency of subpopulations and cell proliferation were found in mice receiving BMC from either group. Also, in the spleen cell number, percentages of B/T cells, B/T-cell proliferation, and interferon γ (IFN-γ) production were similar in all groups. In parallel, a mixture of BMC from congenic sham- and RF-exposed mice were co-transplanted into lymphopenic Rag2 deficient mice. BMC from RF-exposed and sham-exposed mice displayed no advantage or disadvantage when competing for the replenishment of lymphatic organs with mature lymphocytes in Rag2 deficient mice. This model revealed that BMC from sham-exposed and RF-exposed mice were less efficient than BMC from cage control mice in repopulating the thymus, an effect likely due to restraint stress. In conclusion, our results showed no effects of in vivo exposure to GSM-modulated RF-fields on the ability of bone marrow (BM) precursors to long-term reconstitute peripheral T and B cell compartments.


Subject(s)
Bone Marrow Cells/cytology , Bone Marrow Cells/radiation effects , Cell Phone , Electromagnetic Fields/adverse effects , Hematopoiesis/radiation effects , Radio Waves/adverse effects , Animals , Bone Marrow Cells/immunology , Female , Hybridization, Genetic , Mice , Mice, Inbred C57BL , Spleen/immunology , Thymus Gland/immunology , Time Factors
2.
Int J Gynecol Pathol ; 31(6): 507-16, 2012 Nov.
Article in English | MEDLINE | ID: mdl-23018221

ABSTRACT

Two types of ovarian carcinomas are distinguished with respect to morphology, biology, and clinical course, and are designated as Type I and Type II tumors. However, placement of clear cell carcinomas into one of these 2 groups has been problematic as they exhibit morphologic, molecular, and clinical features that do not entirely resemble either Type I or Type II tumors. The present study aimed at better elucidating the clinicopathologic and immunohistochemical features of clear cell carcinomas, in comparison with the 2 main broad categories. To this end, a panel of classic clinicopathologic and immunohistochemical parameters, including estrogen receptor α (ERα), ERß, progesterone receptor, Ki67, p53, and HER2/neu was evaluated in 71 Type I, 157 Type II, and 21 clear cell carcinomas. Overall, findings from the present study support the idea that ovarian clear cell carcinomas are neither Type I nor Type II carcinomas of the ovary; indeed, results obtained showed that similarities between clear cell carcinomas and Type I were limited to the patient's age, tumor dimension, incidence of lymph node and extranodal metastases, and p53 labeling index, whereas the patient's age and incidence of extranodal metastases were the only parameters comparable with the Type II group. The hormonal receptor profile of clear cell carcinomas was characterized by low expression of nuclear ERα and progesterone receptor, and by almost exclusively nuclear ERß immunopositivity, features significantly different from both Type I and II tumors. Finally, the percentage of HER2/neu-positive samples in clear cell carcinomas was 10- and 2.5-fold higher than Type I and Type II ovarian tumors, respectively. In conclusion, our study provides insights into clear cell carcinoma that could help in explaining its unique prognostic features, and, eventually, in orienting toward new therapeutic options.


Subject(s)
Adenocarcinoma, Clear Cell/pathology , Ovarian Neoplasms/pathology , Adenocarcinoma, Clear Cell/chemistry , Female , Humans , Immunohistochemistry , Middle Aged , Neoplasm Staging , Ovarian Neoplasms/chemistry , Receptor, ErbB-2/analysis , Receptors, Estrogen/analysis , Receptors, Progesterone/analysis
3.
Hum Pathol ; 43(2): 282-8, 2012 Feb.
Article in English | MEDLINE | ID: mdl-21835429

ABSTRACT

In this study, we investigated the prognostic value of metastasis tumor antigen 1 expression in 81 untreated patients with ovarian cancer. The expression of metastasis tumor antigen 1 was evaluated by immunohistochemistry, and staining was analyzed in relation to clinicopathologic variables, disease-free survival, and overall survival. High expression of metastasis tumor antigen 1 was found to be associated with advanced stage (I/II versus III/IV, P = .02) and with worse response to first-line treatment (P = .03). Cases with high metastasis tumor antigen 1 expression showed a lower disease-free survival compared with cases with low expression (P = .02). In multivariate analysis of disease-free survival, metastasis tumor antigen 1 overexpression retained an independent negative prognostic role (P = .04), when considered together with histotype, stage of disease, residual tumor at surgery, and chemosensitivity. The evaluation of the prognostic relevance of metastasis tumor antigen 1 in late-stage disease showed that overexpression was a prognostic factor for poor disease-free survival and overall survival in this subset of patients, in both univariate and multivariate models. These findings indicate that metastasis tumor antigen 1 overexpression can be used as a predictor of clinical outcome in patients with ovarian cancer and therefore may represent a new prognostic marker.


Subject(s)
Adenocarcinoma/secondary , Histone Deacetylases/metabolism , Ovarian Neoplasms/pathology , Repressor Proteins/metabolism , Adenocarcinoma/metabolism , Adenocarcinoma/mortality , Adult , Aged , Aged, 80 and over , Biomarkers, Tumor/metabolism , Female , Humans , Middle Aged , Neoplasm Staging , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/mortality , Survival Analysis , Survival Rate , Taiwan/epidemiology , Trans-Activators
4.
Gynecol Oncol ; 122(3): 573-9, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21665249

ABSTRACT

OBJECTIVE: In this study we investigated the prognostic value of estrogen receptor α (ERα), ERß and progesterone receptor (PR) expression in 58 untreated advanced serous ovarian cancer patients. The study also included 12 macroscopically and histopathologically normal ovaries. MATERIALS AND METHODS: Protein expression was evaluated by immunohistochemistry, and antibody staining detected in both the nuclear and cytoplasmic compartments was taken into account. Immunopositivity was analyzed in relation to tumor clinicopathological variables, disease-free survival (DFS), and overall survival (OS). RESULTS: Epithelial cells in ovarian cancer tissue showed significantly lower levels of nuclear ERß and PR, but not ERα, than in normal ovarian tissue. In the case of ERß, however, while normal ovarian epithelium exhibited almost exclusively strong nuclear staining, ovarian cancer tissue mostly showed cytoplasmic immunopositivity. Nuclear ERα and ERß expression were not associated with clinical outcome. Conversely, any cytoplasmic ERß expression was an independent unfavorable prognostic factor for DFS, a finding approaching statistical significance also for OS. These data suggest that, in advanced serous ovarian cancer, cytoplasmic ERß signaling may be more important for patient survival than its nuclear signaling. In the case of PR, positivity was an independent favorable prognostic factor for DFS. CONCLUSIONS: These novel findings, that need to be confirmed in a large prospective trial, suggest that additional prognostic, and possibly therapeutic opportunities may be available in advanced serous ovarian cancer.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cystadenocarcinoma, Serous/drug therapy , Cystadenocarcinoma, Serous/metabolism , Estrogen Receptor beta/biosynthesis , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Adult , Aged , Carboplatin/administration & dosage , Cell Nucleus/metabolism , Cisplatin/administration & dosage , Cohort Studies , Cystadenocarcinoma, Serous/pathology , Cytoplasm/metabolism , Disease-Free Survival , Estrogen Receptor alpha/biosynthesis , Female , Humans , Immunohistochemistry , Middle Aged , Ovarian Neoplasms/pathology , Paclitaxel/administration & dosage , Receptors, Progesterone/biosynthesis
5.
Cancer Lett ; 308(2): 197-202, 2011 Sep 28.
Article in English | MEDLINE | ID: mdl-21636213

ABSTRACT

Gender-related differences in medulloblastoma (MB) development have been reported with a higher incidence in males (slightly above 60%) than in females, female gender being also a significantly favorable prognostic factor in MB. The present study focused on the evaluation of the mechanisms by which estrogens protect against MB formation. To this end, we used a well characterized mouse model of MB - the Patched1 heterozygous mice. Ovariectomized mice were treated with 2,3-bis(4-hydroxyphenyl)-propionitrile (DPN), a highly potent ERß agonist, or 4,4',4″-(4-propyl-[1H]-pyrazole-1,3,5-triyl) trisphenol (PPT), a highly potent ERα agonist. Our results show that the ERß selective agonist DPN significantly inhibits development of MB preneoplastic lesions when compared with untreated ovariectomized mice, restoring the final incidence to that observed in the intact controls, and that these effects were achieved via activation of anti-proliferative and pro-apototic pathways. On the other hand, the ERα selective agonist PPT did not influence MB tumorigenesis relative to untreated ovariectomized mice.


Subject(s)
Estrogen Receptor beta/agonists , Medulloblastoma/pathology , Nitriles/pharmacology , Propionates/pharmacology , Animals , Apoptosis/drug effects , Cell Proliferation/drug effects , Disease Models, Animal , Female , Immunohistochemistry , Male , Mice , Nitriles/therapeutic use , Propionates/therapeutic use , Signal Transduction/drug effects
6.
Cancer Chemother Pharmacol ; 68(1): 107-16, 2011 Jul.
Article in English | MEDLINE | ID: mdl-20848284

ABSTRACT

PURPOSE: Hyaluronan (HA)-receptors (mainly CD44 and RHAMM) are overexpressed in a wide variety of cancers including ovarian tumors, and HA-bioconjugates have been developed to enhance selective entry of cytotoxic drugs into HA receptor-expressing cancerous cells. Here, we evaluated the potential application of a new HA-paclitaxel bioconjugate, ONCOFID-P, for intraperitoneal (IP) treatment of ovarian cancer. METHODS: In vitro cytotoxic effect of ONCOFID-P was first assessed on CD44(+) OVCAR-3 and SKOV-3 human ovarian cancer cell lines. Studies were performed in female Balb/c athymic mice IP implanted with OVCAR-3 or SKOV-3 and treated with IP ONCOFID-P, and IP and intravenous (IV) free paclitaxel, at their maximum tolerated dose (MTD 168, 80 and 80 mg/kg, total dose, respectively). The potential detrimental effect of the IP ONCOFID-P and IP free paclitaxel on hematopoiesis was also assessed on peripheral blood, bone marrow and spleen. RESULTS: Results show that ONCOFID-P cytotoxicity against both OVCAR-3 and SKOV-3 cell lines was somewhat less effective than free paclitaxel. Conversely, in in vivo experiments, IP treatment with ONCOFID-P was overall more effective than IV and IP free paclitaxel in inhibiting intra-abdominal tumor dissemination, abrogating ascites, prolonging survival and curing mice. ONCOFID-P and IP free paclitaxel were equivalent in terms of myelotoxicity, although the former was administered at a two-fold higher dose. CONCLUSIONS: Present data strongly support the development of ONCOFID-P for locoregional treatment of ovarian cancer.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Hyaluronic Acid/analogs & derivatives , Hyaluronic Acid/therapeutic use , Ovarian Neoplasms/drug therapy , Paclitaxel/analogs & derivatives , Paclitaxel/therapeutic use , Animals , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/toxicity , CA-125 Antigen/metabolism , Cell Line, Tumor , Female , Humans , Hyaluronan Receptors/metabolism , Hyaluronic Acid/administration & dosage , Hyaluronic Acid/toxicity , Injections, Intraperitoneal , Mice , Mice, Inbred BALB C , Ovarian Neoplasms/immunology , Ovarian Neoplasms/pathology , Paclitaxel/administration & dosage , Paclitaxel/toxicity , Treatment Outcome , Xenograft Model Antitumor Assays
7.
Transl Oncol ; 3(5): 326-35, 2010 Oct 01.
Article in English | MEDLINE | ID: mdl-20885895

ABSTRACT

Medulloblastoma is the most frequent malignant pediatric brain tumor with a dismal prognosis in 30% of cases. We examined the activity of AEE788, a dual inhibitor of human epidermal receptor (HER) 1/2 and vascular endothelial growth factor receptor (VEGFR) 1/2, in medulloblastoma preclinical models. Established lines (Daoy and D283), chemoresistant (Daoy(Pt)), and ectopically HER2-overexpressing (Daoy(HER2)) cells expressed diverse levels of total and activated AEE788 target receptors. In vitro, AEE788 inhibited cell proliferation (IC(50) from 1.7 to 3.8 µM) and prevented epidermal growth factor- and neuregulin-induced HER1, HER2, and HER3 activation. Inhibition of Akt paralleled that of HER receptors. In vivo, AEE788 growth inhibited Daoy, Daoy(Pt), and Daoy(HER2) xenografts by 51%, 45%, and 72%, respectively. Immunohistochemical analysis of mock- and HER2-transfected xenografts revealed that the latter showed, along with high HER2 expression, high VEGFR2 staining in tumor and endothelial cells and increased expression of the endothelial marker CD31. AEE788 reduced the activation of target receptors and angiogenesis. In 21 primary medulloblastoma, HER2 expression significantly correlated (P < .01) with VEGFR2 (r = 0.56) and VEGF (r = 0.61). In conclusion, AEE788 shows similar growth-suppressive activities in chemosensitive and chemoresistant medulloblastoma cells in vitro and in vivo. Ectopic HER2 overexpression sensitizes cells to AEE788 in vivo, but not in vitro, possibly through host-mediated processes. Together with the experimental data, the finding that HER2 positively correlates with VEGFR2 and VEGF in human medulloblastoma specimens indicates HER2-overexpressing medulloblastoma as the subset that most likely might benefit from AEE788 treatment.

8.
Virchows Arch ; 457(1): 27-34, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20552366

ABSTRACT

This study was aimed at determining whether high-grade endometrioid carcinomas (grade 3 International Federation of Gynecology and Obstetrics) might overlap, at least partially, non-endometrioid carcinomas (type II). To this end, a panel of clinical-pathological and immunohistochemical parameters was evaluated in three different populations: low-grade endometrioid carcinomas (LGECs; n = 57), high-grade endometrioid carcinomas (HGECs; n = 26), and non-endometrioid carcinomas (NECs; n = 30). Besides morphological appearance, HGECs appeared similar to LGECs in p53 immunostaining profile; features different from LGECs included a higher local aggressiveness, a higher invasion of lymph-vascular spaces, a lower expression of ERalpha and PR, and a higher proliferative index. HGECs were similar to NECs for local aggressiveness, invasion rate of lymph-vascular spaces, lymph node metastasis incidence, and proliferative index. HGECs, however, showed a lower rate of extra-nodal metastases, a lower incidence of p53 overexpression, and a higher positivity for ERalpha and PR. In conclusion, results from this study show that HGECs exhibit overlapping morphological and immunohistochemical features of both type I and type II endometrial carcinomas. Further research is needed to clarify the clinical value of this observation.


Subject(s)
Carcinoma, Endometrioid/classification , Carcinoma, Endometrioid/pathology , Endometrial Neoplasms/classification , Endometrial Neoplasms/pathology , Biomarkers, Tumor/metabolism , Carcinoma, Endometrioid/metabolism , Endometrial Neoplasms/metabolism , Female , Humans , Immunohistochemistry , Ki-67 Antigen , Middle Aged , Neoplasm Staging , Receptors, Estrogen/biosynthesis , Receptors, Progesterone/biosynthesis , Tumor Suppressor Protein p53
9.
Int J Cancer ; 127(12): 2749-57, 2010 Dec 15.
Article in English | MEDLINE | ID: mdl-21351254

ABSTRACT

Medulloblastoma (MB) is the most common pediatric tumor of the CNS, representing ∼20% of all childhood CNS tumors. Although in recent years many molecular mechanisms that control MB development have been clarified, the effects of biological factors such as sex on this tumor remain to be explained. Epidemiological data, in fact, indicate a significant difference in the incidence of MB between the 2 sexes, with considerably higher susceptibility of males than females. Besides this different susceptibility, female sex is also a significant favorable prognostic factor in MB, with girls having a much better outcome. Despite these literature data, there has been little investigation into estrogen influence on MB development. In our study, we evaluated how hormone deficiency resulting from ovariectomy and hormone replacement influences the development of early and advanced MB stages in Patched1 heterozygous mice, a well-characterized mouse model of radiation-induced MB. Susceptibility to MB development was significantly increased in ovariectomized Ptch1(+/-) females and restored to levels observed in control mice after estrogen replacement. We next investigated the molecular mechanisms by which estrogen might influence tumor progression and show that ERß, but not ERα, is involved in modulation of MB development by estrogens. Finally, our study shows that a functional interaction between estrogen- and IGF-I-mediated pathways may be responsible for the effects observed.


Subject(s)
Cerebellar Neoplasms/prevention & control , Estradiol/therapeutic use , Estrogens/therapeutic use , Medulloblastoma/prevention & control , Receptors, Cell Surface/physiology , Animals , Blotting, Western , Cerebellar Neoplasms/genetics , Cerebellar Neoplasms/pathology , Estrogen Receptor beta/metabolism , Female , Heterozygote , Immunoenzyme Techniques , Insulin-Like Growth Factor I/metabolism , Medulloblastoma/genetics , Medulloblastoma/pathology , Mice , Mice, Knockout , Ovariectomy , Patched Receptors , Patched-1 Receptor , RNA, Messenger/genetics , Receptors, Estrogen/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Whole-Body Irradiation
10.
Biochem Pharmacol ; 78(11): 1374-81, 2009 Dec 01.
Article in English | MEDLINE | ID: mdl-19643088

ABSTRACT

This study was aimed at evaluating the potential application of benzophenanthridine alkaloids, sanguinarine and cheleritrine, in the therapy of melanoma cancer. In vitro antiproliferative activity of sanguinarine was higher than that of cheleritrine against the B16 melanoma 4A5 cells. Both agents were able to produce DNA breaks, and the DNA unwinding assay showed that they act as DNA intercalating agents. Sanguinarine was selected for determination of its in vivo preclinical efficacy. Oral treatment with sanguinarine reduced the tumor burden in a transplantable murine tumor grown in a syngeneic host (B16 melanoma 4A5 in C57BL/6 mice), and in a human tumor xenograft grown in immunodeficient mice (A375 human melanoma in athymic nude mice). In A375 tumors a significant decrease in the proliferation marker Ki67, and a reduction in the activated mitogen-activated protein kinases (p-p44/42 MAPK), and in protein kinase B (pAKT) were also observed. Three out of eleven A375-bearing treated mice were tumor-free at the end of treatment, and did not develop any tumor after a further, treatment-free, observation period of 60 days. Sanguinarine also showed a striking antiangiogenic activity in mice. Data from the present study support the concept that sanguinarine can be effective in melanoma skin cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Benzophenanthridines/pharmacology , Isoquinolines/pharmacology , Melanoma, Experimental/drug therapy , Angiogenesis Inhibitors/pharmacology , Angiogenesis Inhibitors/therapeutic use , Animals , Antineoplastic Agents/therapeutic use , Benzophenanthridines/therapeutic use , Cell Line, Tumor , Cell Proliferation/drug effects , Collagen , Drug Combinations , Female , Humans , Isoquinolines/therapeutic use , Laminin , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Nude , Neoplasm Transplantation , Proteoglycans , Transplantation, Heterologous
11.
BMC Cancer ; 9: 221, 2009 Jul 07.
Article in English | MEDLINE | ID: mdl-19583859

ABSTRACT

BACKGROUND: Much attention has been recently focused on the role of cancer stem cells (CSCs) in the initiation and progression of solid malignancies. Since CSCs are able to proliferate and self-renew extensively, thus sustaining tumor growth, the identification of CSCs through their antigenic profile might have relevant clinical implications. In this context, CD133 antigen has proved to be a marker of tumor cells with stemness features in several human malignancies.The aim of the study was to investigate the clinical role of the immunohistochemically assessed expression of CD133 in a large single Institution series of ovarian cancer patients. METHODS: The study included 160 cases admitted to the Gynecologic Oncology Unit, Catholic University of Campobasso and Rome. CD133 antigen was identified by the monoclonal mouse anti-CD133-1 antibody (clone CD133 Miltenyi biotec). RESULTS: In the overall series CD133 positive tumor cells were observed in 50/160 (31.2%) cases. A diffuse cytoplasmic pattern was identified in 30/50 (60.0%), while an apical cytoplasmic pattern was found in 20/50 (40.0%) of CD133 positive tumors.As of September 2008, the median follow up was 37 months (range: 2-112). During the follow up period, progression and death of disease were observed in 123 (76.9%), and 88 (55.0%) cases, respectively. There was no difference in TTP between cases with negative (median TTP = 23 months) versus positive CD133 expression (median TTP = 24 months) (p value = 0.3). Similar results were obtained for OS. When considering the TTP and OS curves according to the pattern of CD133 expression, a trend to a worse prognosis for cases with diffuse cytoplasmic versus the apical cytoplasmic pattern was documented, although the statistical significance was not reached. CONCLUSION: The immunohistochemical assessment of CD133 expression seems not to provide additional prognostic information in ovarian cancer patients. The role of the different pattern of CD133 immunoreaction deserves further investigation in a larger series.


Subject(s)
Antigens, CD/biosynthesis , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Glycoproteins/biosynthesis , Ovarian Neoplasms/metabolism , AC133 Antigen , Aged , Animals , Antibodies, Monoclonal/chemistry , Area Under Curve , Disease Progression , Female , Humans , Immunohistochemistry/methods , Mice , Middle Aged , Peptides , Time Factors
12.
Clin Cancer Res ; 15(13): 4299-311, 2009 Jul 01.
Article in English | MEDLINE | ID: mdl-19509143

ABSTRACT

PURPOSE: Cancer stem cells represent an attractive therapeutic target for tumor eradication. The present study aimed to determine whether CD133 expression may identify cells with characteristics of cancer stem/progenitor cells in human endometrial tumors. EXPERIMENTAL DESIGN: We analyzed 113 tumor samples for CD133/1 expression by flow cytometry, immunohistochemistry, and semiquantitative reverse transcription-PCR. CD133(+) cells were isolated and used to assess phenotypic characteristics, self-renewal capacity, ability to maintain CD133 expression and form sphere-like structures in long-term cultures, sensitivity to chemotherapeutic agents, gene expression profile, and ability to initiate tumors in NOD/SCID mice. RESULTS: Primary tumor samples exhibited a variable degree of immunoreactivity for CD133/1, ranging from 1.3% to 62.6%, but stained negatively for other endothelial and stem cell-associated markers. Isolated CD133(+) cells expanded up to 4.6-fold in serum-replenished cultures and coexpressed the GalNAcalpha1-O-Ser/Thr MUC-1 glycoform, a well-characterized tumor-associated antigen. Dissociated bulk tumors formed sphere-like structures; cells grown as tumor spheres maintained CD133 expression and could be propagated for up to 12 weeks. CD133(+) cells purified from endometrioid adenocarcinomas were resistant to cisplatin-induced and paclitaxel-induced cytotoxicity and expressed a peculiar gene signature consisting of high levels of matrix metalloproteases, interleukin-8, CD44, and CXCR4. When serially transplanted into NOD/SCID mice, CD133(+) cells were capable of initiating tumor formation and recapitulating the phenotype of the original tumor. CONCLUSIONS: CD133 is expressed by human endometrial cancers and might represent a valuable tool to identify cells with cancer stem cell characteristics.


Subject(s)
Antigens, CD/metabolism , Endometrial Neoplasms/pathology , Endometrium/pathology , Glycoproteins/metabolism , Neoplastic Stem Cells/metabolism , Peptides/metabolism , AC133 Antigen , Adult , Aged , Aged, 80 and over , Animals , Biomarkers, Tumor/analysis , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Carcinoma/diagnosis , Carcinoma/genetics , Carcinoma/metabolism , Carcinoma/pathology , Endometrial Neoplasms/diagnosis , Endometrial Neoplasms/genetics , Endometrial Neoplasms/metabolism , Endometrium/metabolism , Female , Gene Expression Profiling , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Middle Aged , Neoplastic Stem Cells/pathology , Oligonucleotide Array Sequence Analysis , Transplantation, Heterologous , Tumor Cells, Cultured
13.
Gynecol Oncol ; 111(2): 237-43, 2008 Nov.
Article in English | MEDLINE | ID: mdl-18718649

ABSTRACT

OBJECTIVE: The aim of the study was to investigate the potential clinical relevance of immunohistochemically assessed RON expression in a large, single institution series of primary untreated advanced ovarian cancer patients. METHODS: Immunohistochemical analysis was performed by using the polyclonal rabbit anti-RON-beta antibody (C-20, clone sc-322, Santa Cruz, California). Results were expressed as the total proportion of immunostained tumor cells (RON positivity), or the percentage of cells showing strong staining of RON expression (H-RON positivity). RESULTS: In the overall series RON positive immunoreaction was observed in 103/141 cases, while H-Ron positivity was detected in 577141 (40.4%) cases. No association between RON and H-RON expression with response to first-line treatment was documented. During the follow up period, progression and death of disease were observed in 111 (78.7%) and 76 (53.9%) cases, respectively. Cases with strong H-RON expression has a shorter overall survival (median=35 months) than cases with low RON levels (median=59 months) (X(2)=-2.1, p value=0.032). In multivariate analysis, only platinum resistance, and extent of residual tumor retained an independent negative prognostic role for OS, with the percentages of H-RON positively immunostained cells showing a borderline statistical significance (p value=0.0643). The unfavourable role of elevated percentages of H-RON expression was maintained only in the subgroup of platinum resistant recurrent ovarian cancer patients (X(2)=3.89, p value=0.048) compared to the platinum sensitive ones (X(2)=1.98, p value=0.16). CONCLUSIONS: The assessment of RON expression deserves further attention as a parameter helpful to identify poor prognosis ovarian cancer patients potentially candidates to investigational agents.


Subject(s)
Ovarian Neoplasms/metabolism , Receptor Protein-Tyrosine Kinases/biosynthesis , Adult , Aged , Aged, 80 and over , Female , Humans , Immunohistochemistry , Middle Aged , Ovarian Neoplasms/pathology , Prognosis , Survival Rate , Young Adult
14.
Radiat Res ; 170(6): 803-10, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19138032

ABSTRACT

We examined the effects of in vivo exposure to a GSM-modulated 900 MHz RF field on the ability of bone marrow cells to differentiate, colonize lymphatic organs, and rescue lethally X-irradiated mice from death. X-irradiated mice were injected with medium alone or containing bone marrow cells from either RF-field-exposed (SAR 2 W/kg, 2 h/day, 5 days/ week, 4 weeks) or sham-exposed or cage control donor mice. Whereas all mice injected with medium alone died, mice that received bone marrow cells survived. Three and 6 weeks after bone marrow cell transplantation, no differences in thymus cellularity and in the frequencies of differentiating cell subpopulations (identified by CD4/CD8 expression) were observed among the three transplanted groups. Mitogen-induced thymocyte proliferation yielded comparable levels in all transplanted groups. As to the spleen, no effects of the RF-field exposure on cell number, percentages of B and T (CD4 and CD8) cells, B- and T-cell proliferation, and IFN-gamma production were found in transplanted mice. In conclusion, our results show no effect of in vivo exposure to GSM-modulated RF fields on the ability of bone marrow precursor cells to home and colonize lymphoid organs and differentiate in phenotypically and functionally mature T and B lymphocytes.


Subject(s)
Bone Marrow Cells/radiation effects , Electromagnetic Fields , Radio Waves , Animals , B-Lymphocytes/cytology , B-Lymphocytes/radiation effects , Bone Marrow Cells/metabolism , Bone Marrow Transplantation , Cell Count , Cell Differentiation/radiation effects , Cell Proliferation/radiation effects , Female , Mice , Spleen/cytology , Spleen/radiation effects , Survival Rate , T-Lymphocytes/cytology , T-Lymphocytes/radiation effects , Thymus Gland/cytology , Thymus Gland/radiation effects
15.
Radiat Res ; 165(6): 664-70, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16802866

ABSTRACT

We examined the effects of in vivo exposure to a GSM-modulated 900 MHz RF field on B-cell peripheral differentiation and antibody production in mice. Our results show that exposure to a whole-body average specific absorption rate (SAR) of 2 W/kg, 2 h/day for 4 consecutive weeks does not affect the frequencies of differentiating transitional 1 (T1) and T2 B cells or those of mature follicular B and marginal zone B cells in the spleen. IgM and IgG serum levels are also not significantly different among exposed, sham-exposed and control mice. B cells from these mice, challenged in vitro with LPS, produce comparable amounts of IgM and IgG. Moreover, exposure of immunized mice to RF fields does not change the antigen-specific antibody serum level. Interestingly, not only the production of antigen-specific IgM but also that of IgG (which requires T-B-cell interaction) is not affected by RF-field exposure. This indicates that the exposure does not alter an ongoing in vivo antigen-specific immune response. In conclusion, our results do not indicate any effects of GSM-modulated RF radiation on the B-cell peripheral compartment and antibody production and thus provide no support for health-threatening effects.


Subject(s)
Antibody Formation/physiology , B-Lymphocytes/cytology , B-Lymphocytes/immunology , Cell Phone , Electromagnetic Fields , Microwaves , Whole-Body Irradiation , Animals , Antibody Formation/radiation effects , B-Lymphocytes/radiation effects , Cell Differentiation/radiation effects , Cells, Cultured , Dose-Response Relationship, Radiation , Female , Mice , Mice, Inbred C57BL , Radiation Dosage
SELECTION OF CITATIONS
SEARCH DETAIL
...